WO2013128381A1 - Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection - Google Patents

Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection Download PDF

Info

Publication number
WO2013128381A1
WO2013128381A1 PCT/IB2013/051562 IB2013051562W WO2013128381A1 WO 2013128381 A1 WO2013128381 A1 WO 2013128381A1 IB 2013051562 W IB2013051562 W IB 2013051562W WO 2013128381 A1 WO2013128381 A1 WO 2013128381A1
Authority
WO
WIPO (PCT)
Prior art keywords
aat
temporary
transplantation
cell
graft
Prior art date
Application number
PCT/IB2013/051562
Other languages
English (en)
French (fr)
Inventor
Eli Lewis
Original Assignee
Ben Gurion University Of The Negev Research And Development Authority
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ben Gurion University Of The Negev Research And Development Authority filed Critical Ben Gurion University Of The Negev Research And Development Authority
Priority to US14/380,118 priority Critical patent/US20150010581A1/en
Priority to EP13755285.7A priority patent/EP2819698A4/de
Publication of WO2013128381A1 publication Critical patent/WO2013128381A1/en
Priority to IL233647A priority patent/IL233647A0/en
Priority to US15/477,379 priority patent/US10086070B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/39Pancreas; Islets of Langerhans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/14Peptides containing saccharide radicals; Derivatives thereof, e.g. bleomycin, phleomycin, muramylpeptides or vancomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/217IFN-gamma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to compositions and methods for the prevention and treatment of graft rejection, including xenograft rejection, and for attenuating host responses in transplantation of cells, pancreatic islets, tissues and organs. More specifically, the compositions and methods of the present invention relate to combined therapies comprising treatment of alpha- 1- antitrypsin and temporary T-cell depletion in the graft recipient.
  • Transplantation systems such as organ transplantations have become important, effective and at times the sole therapies for many life-threatening end- stage diseases.
  • injurious immune responses are still the major barrier for successful transplantation. This is manifested in irreversible and life-threatening graft failure (host- versus-graft response, or HVG) or pathological immune reactivity of bone-marrow transplants graft-versus-host disease (GVHD).
  • HVG host- versus-graft response
  • GVHD pathological immune reactivity of bone-marrow transplants graft-versus-host disease
  • Pancreatic islet transplantation can provide type-1 diabetes patients with functional islets and physiological circulating glucose levels.
  • shortage of human donors represents a critical obstacle.
  • Islet xenograft transplantation from non-human donors provides an alternative for human islet allotransplantation; in addition to providing an array of islet sources, xenografts offer the advantage of elective procedures (that is, the donor is recruited upon availability rather than the patient), and potentially manipulating donor cells towards superior islet function.
  • elective procedures that is, the donor is recruited upon availability rather than the patient
  • Xenograft rejection is largely attributed to vast antigen disparity between species, thus triggering multiple arms of the immune response. Indeed, in addition to host CD4 + T cell involvement, evidence suggests that CD8 + T cells and B cells partake in xenograft rejection. Additionally, inflammation limits islet xenograft survival, particularly in early days post-transplantation, a challenging therapeutic obstacle considering that diabetogenic corticosteroids are excluded from current islet transplantation protocols. Within this context, the desired emergence of protective regulatory T cells (Tregs) appears further intangible.
  • T cell debulking therapy a regimen comprised of polyclonal antibodies that temporarily deplete T cells, is currently used for prevention of acute rejection in organ transplantation.
  • Combination of anti-CD4 and anti-CD8 antibodies in mice may represent the use of ATG in patients, as it achieves a similar temporary decline in T-cell numbers (Tchorsh-Yutsis et al. Transplantation 2011;91(4):398-405; Tchorsh-Yutsis et al.
  • hAAT Human a 1 -antitrypsin
  • US Pat. Appl. No. 20090220518 to an inventor of the current invention and co- workers, relates to treating, reducing or preventing transplantation rejection and/or side effects associated with transplantation.
  • xenograft rejection may be prevented by combination therapy comprising AAT and temporary T cell depletion, particularly, anti-CD4 and anti-CD8 antibodies administration.
  • the present invention provides a method of preventing or treating xenotransplant rejection in a subject in need thereof, the method comprises administering to said subject a therapeutically effective amount of AAT in combination with a therapeutically effective amount of at least one temporary T cell depleting agent.
  • the at least one temporary T cell depleting agent is selected from anti-CD4 and anti-CD8 antibodies, or an antigen binding fragment thereof.
  • the at least one temporary T cell depleting agent is an anti-CD4 antibody, or an antigen binding fragments thereof.
  • the at least one temporary T cell depleting agent is an anti-CD8 antibody, or an antigen binding fragment thereof.
  • the at least one temporary T cell depleting agent is anti-CD4 and anti-CD8 antibodies, or antigen binding fragments thereof.
  • said graft is selected from the group consisting of pancreatic islet cells, hematopoietic cells, stem cells, pancreas, heart, lung, kidney, liver or skin.
  • the graft is pancreatic islet cells.
  • the graft is hematopoietic cell.
  • said graft is a xenograft.
  • Figure 1 depicts human AAT monotherapy during pancreatic islet xenotransplantation.
  • Rat pancreatic islets were grafted into the renal subcapsular space of hyperglycemic mice. Recipients were treated with saline (CT) or human AAT throughout the experiment.
  • CT saline
  • B Graft histology.
  • Figure 2 is a graphic illustration of draining lymph nodes (DLN) response to human AAT monotherapy after skin xenografting. Mice were either SHAM operated (CT) or recipients of rat skin (Tx) in the absence or presence of human AAT monotherapy.
  • CT SHAM operated
  • Tx rat skin
  • (A) 14- day DLN. FACS analysis. Results expressed as fold change from CT, mean + SEM from n 10/group; ** p ⁇ 0.01, *** p ⁇ 0.001.
  • Figure 3 depicts graft survival following AAT treatment combined with debulking therapy.
  • A CD45 + CD3 + cells from peripheral blood, as monitored by FACS analysis. Results presented as the percent out of initial amount prior to injection. Representative follow-up out of 10 mice.
  • B Islet xenograft survival curve. *** p ⁇ 0.001 between DB and BD/AAT.
  • C The percentage of mice having functional islet xenograft following CT, DB/AAT, anti-CD8, and anti-CD8/AAT treatments.
  • mice having functional islet xenograft following CT, DB/AAT, anti-CD4, and anti-CD4/AAT treatments.
  • E Glucose follow-up. Representative mouse. Milestones indicated: hAAT treatment stopped, therapy withdrawn followed by glucose follow-up; nephrectomy, graft explantation followed by glucose follow-up; second xenograft, rat islets grafted into the right renal subcapsular space followed by glucose follow-up.
  • FIG 4 illustrates AAT treatment combined with debulking therapy; histology and gene expression.
  • Figure 5 are graphs showing that hAAT promotes expansion of foxp3 positive CD4 T-cells and delays CD8 T-cell re-population after T-cell depletion.
  • C57BL/6 (WT) and hAAT transgenic mice (hAAT +/+ ) n 5 per group underwent systemic T-cell depletion using the combination of anti-CD4 and anti-CD8 depleting antibodies.
  • (A) shows the interplay between CD3 and CD45 expression;
  • B) shows the interplay between CD8 and CD3 expression;
  • C shows the interplay between CD4 and CD3 expression;
  • B shows the interplay between F0xp3 and CD4 expression.
  • the invention is directed to compositions and methods for the prevention and treatment of xenograft rejection, and for attenuating host immune responses following xenograft transplantation of tissues, organs and cells. Further, the present invention provides compositions and methods for suppressing the immune response of a graft recipient non-responsive or resistant to a first line treatment, including, but not limited to subjects afflicted with graft dysfunction.
  • hAAT Human AAT
  • hAAT monotherapy has been recently shown to protect islet allografts from acute rejection and facilitates strain- specific immune tolerance, however, hAAT monotherapy appears insufficient to allow xenograft acceptance.
  • AAT monotherapy resulted in xenografts rejection despite attempts to prolong the treatment and/or extend its time course. Considering that xenograft rejection is difficult to control, this would seem the final option for involvement of AAT in this context.
  • an attempt to combine AAT therapy with co- stimulation blockade using anti-CD 154/LFA-l did not result in significant change in xenotransplant rejection as well.
  • a subject afflicted with graft dysfunction refers to the earliest point of detection of an ongoing graft's failure.
  • a subject afflicted with graft dysfunction is, in some embodiment, a graft recipient non-responsive to first-line immunosuppressive protocol or, in additional embodiments, any subsequent immunosuppressive treatment.
  • said subject is a treatment-resistant subject.
  • said first-line immunosuppressive treatment is steroid treatment, including but not limited to corticosteroids.
  • Corticosteroid therapy is typically administered at a high dose at the time of transplantation and then gradually reduced to a maintenance dose, which is given indefinitely. The approach ablates immune responses, but does not alter the profile of the immune cells that recover from the effects of steroids.
  • the transplanted cells are genetically modified cells.
  • genetically modified cells as referred to herein relates to cells being transfected by a vector, as exemplified by an expression vector comprising the coding sequence of a gene of interest, said cells capable of expressing said gene.
  • Methods for genetically modifying cells such as hematopoietic cells, stem cells or pancreatic islet cells are well known in the art.
  • therapeutically effective amounts is intended to qualify the amount of each agent for use in the combination therapy which will achieve the goal of improvement in severity and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative therapies.
  • the therapeutically effective amount of at least one agent of the invention (AAT or T cell depleting agent) is lower than the amount used in monotherapy using said agent.
  • the therapeutically effective amount of AAT is lower than the amount used in monotherapy using said agent.
  • AAT is administered at a dose of 5-300 mg/kg. According to some embodiments, AAT is administered at a dose of 10-280 mg/kg. According to some embodiments, AAT is administered at a dose of 15-260 mg/kg. According to another embodiment, AAT is administered at a dose of 45-240 mg/kg.
  • the anti-CD4 antibody is GK1.5. In another embodiment, the anti-CD8 antibody is 53.6.72. Said antibodies are commercially available such as from BioXCell. In another embodiment, the anti-CD4 antibody exhibits similar T cell depleting activity as the GK1.5 antibody. In another embodiment, the anti-CD8 antibody exhibits similar T cell depleting activity as the 53.6.72 antibody.
  • T cell depleting agents are known to one skilled in the art. Non limiting examples for T cell depleting agents include anti-CD3, anti-CD4, anti-CD25, anti-CD8, anti-CD8a, anti-TCR, anti-TCR-gamma-delta and anti-thymocyte-globulin (ATG). Each possibility is a separate embodiment of the present invention.
  • AAT administration is a long term administration.
  • said AAT administration is selected from single-dose administration or sequential administration.
  • AAT is administered prior to transplantation, following transplantation or a combination thereof.
  • administering AAT prior to treatment is for no more than 10 days prior to transplantation.
  • the AAT is human AAT (hAAT).
  • said hAAT comprises an amino acid sequence as set forth in SEQ ID NO: 1.
  • said hAAT consists of an amino acid sequence as set forth in SEQ ID NO: 1
  • substitutions, deletions or additions to a peptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a conservatively modified variant where the alteration results in the substitution of an amino acid with a similar charge, size, and/or hydrophobicity characteristics, such as, for example, substitution of a glutamic acid (E) to aspartic acid (D).
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • analog includes any peptide having an amino acid sequence substantially identical to one of the sequences specifically shown herein in which one or more residues have been conservatively substituted with a functionally similar residue and which displays the abilities as described herein.
  • conservative substitutions include the substitution of one non-polar (hydrophobic) residue such as isoleucine, valine, leucine or methionine for another, the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, between glycine and serine, the substitution of one basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue, such as aspartic acid or glutamic acid for another.
  • a non-polar (hydrophobic) residue such as isoleucine, valine, leucine or methionine for another
  • one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and
  • a peptide derived from hAAT can be an analog, fragment, conjugate (e.g. a lipopeptide conjugate) or derivative of a native hAAT, and salts thereof, as long as said peptide retains its ability to protect the transplant from inflammation.
  • the present invention encompasses derivatives of AAT.
  • derivatives or “chemical derivative” includes any chemical derivative of AAT having one or more residues chemically derivatized by reaction of side chains or functional groups.
  • chemical derivatives are those peptides, which contain one or more naturally occurring amino acid derivatives of the twenty standard amino acid residues.
  • 4-hydroxyproline may be substituted for proline
  • 5-hydroxylysine may be substituted for lysine
  • 3-methylhistidine may be substituted for histidine
  • homoserine may be substituted or serine
  • ornithine may be substituted for lysine.
  • a derivative can differ from the natural sequence of the peptides of the invention by chemical modifications including, but are not limited to, terminal-NH 2 acylation, acetylation, or thioglycolic acid amidation, and by terminal-carboxlyamidation, e.g., with ammonia, methylamine, and the like.
  • Peptides can be either linear, cyclic or branched and the like, which conformations can be achieved using methods well known in the art.
  • the present invention also encompasses derivatives and analogs in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonylamino groups, carbobenzoxyamino groups, t-butyloxycarbonylamino groups, chloroacetylamino groups or formylamino groups.
  • Free carboxyl groups may be derivatized to form, for example, salts, methyl and ethyl esters or other types of esters or hydrazides.
  • the imidazole nitrogen of histidine can be derivatized to form N-im-benzylhistidine.
  • the analogs can also contain non-natural amino acids.
  • non-natural amino acids include, but are not limited to, sarcosine (Sar), norleucine, ornithine, citrulline, diaminobutyric acid, homoserine, isopropyl Lys, 3-(2'-naphtyl)-Ala, nicotinyl Lys, amino isobutyric acid, and 3-(3'-pyridyl-Ala).
  • Specific examples include, but are not limited to, methyl-Ala (Me Ala), MeTyr, MeArg, MeGlu, MeVal, MeHis, N-acetyl-Lys, O- acetyl-Lys, carbobenzoxy-Lys, Tyr-O-Benzyl, Glu-O-Benzyl, Benzyl-His, Arg-Tosyl, t- butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, and the like.
  • compositions of the invention can be formulated in the form of a pharmaceutically acceptable salt of the peptides of the present invention or their analogs or derivatives thereof.
  • Pharmaceutically acceptable salts include those salts formed with free amino groups such as salts derived from non-toxic inorganic or organic acids such as hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those salts formed with free carboxyl groups such as salts derived from non-toxic inorganic or organic bases such as sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like.
  • the composition can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates or phosphates.
  • Antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; and agents for the adjustment of tonicity such as sodium chloride or dextrose are also envisioned.
  • compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, gels, creams, ointments, foams, pastes, sustained-release formulations and the like.
  • the compositions can be formulated as a suppository, with traditional binders and carriers such as triglycerides, microcrystalline cellulose, gum tragacanth or gelatin.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in: Remington's Pharmaceutical Sciences by E.W. Martin, the contents of which are hereby incorporated by reference herein.
  • Toxicity and therapeutic efficacy of the compositions described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the IC50 (the concentration which provides 50% inhibition) and the LD50 (lethal dose causing death in 50 % of the tested animals) for a subject compound.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See, for example, Fingl et al., 1975, in The Pharmacological Basis of Therapeutics, Ch. 1 p.l, the contents of which are hereby incorporated by reference in their entirety).
  • dosing can also be a single administration of a slow release composition, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions of the present invention can be supplied in any manner suitable for the provision of the peptide to cells within the tissue of interest.
  • a composition of the present invention can be introduced, for example, into the systemic circulation, which will distribute the peptide to the tissue of interest.
  • a composition can be applied topically to the tissue of interest (e.g., injected, or pumped as a continuous infusion, or as a bolus within a tissue, applied to all or a portion of the surface of the skin, etc.).
  • Suitable routes of administration include, but are not limited to, parenteral injections, e.g., intradermal, intravenous, intramuscular, intralesional, subcutaneous, intrathecal, and any other mode of injection as known in the art.
  • parenteral injections e.g., intradermal, intravenous, intramuscular, intralesional, subcutaneous, intrathecal, and any other mode of injection as known in the art.
  • bioavailability of polypeptides administered by other routes can be lower than when administered via parenteral injection, by using appropriate formulations it is envisaged that it will be possible to administer the compositions of the invention via transdermal, oral, rectal, vaginal, topical, nasal, inhalation and ocular modes of treatment.
  • the pharmaceutical composition may be in the form of tablets or capsules, which can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose; a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; or a glidant such as colloidal silicon dioxide.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose
  • a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate
  • a glidant such as colloidal silicon dioxide.
  • dosage unit form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • dosage unit forms can contain various other materials which modify
  • hAAT lung-specific transgenic mice (C57BL/6 background) were a kind gift from Prof. A. Churg (University of British Columbia, Vancouver, Canada). Six-to- eight-week old heterozygote siblings from breeding couples of WT C57BL/6 (Harlan laboratories Inc., Israel) x human AAT lung-specific transgenic mice were used as graft recipients, as described elsewhere (19). Nine-to-ten- week old Sprague Dawley female rats (Harlan laboratories) were used as pancreatic islet and skin donors. Experiments were approved by institutional Animal Care and Use Committee.
  • Pancreatic islet isolation Donor rats were anesthetized and then bled. The bile duct was ligated at the liver and at the intestinal ends, then cannulated with a 27G needle. The pancreas was inflated with 10 ml cold collagenase (1 mg/ml, type XI, Sigma, Israel), removed and incubated for 17 minutes at 37 °C while continuously stirred with a 3 mm sterile magnet. Digested pancreas was mechanically sheared by vortex and tissue was filtered through a 1,000 ⁇ sieve. Islets were collected from a double-Ficoll gradient (1.0771 and 1.1191, Sigma).
  • HBSS Hanks balanced salt solution
  • BSA bovine serum albumin
  • FCS fetal calf serum
  • pancreatic islets were then hand-picked under a stereoscope into a culture flask and incubated overnight.
  • Islet xenotransplantation Islet transplantation in the renal subcapsular space was performed as described, with minor modifications (19). Rat islets (315-400/transplant) were implanted under the renal capsule of recipient mice that were rendered hyperglycemic by single-dose streptozotocin (225 mg/kg, Sigma). A relatively small number of xenogeneic islets (315-400) were implanted. Prospective recipients were screened for non-fasting circulating glucose levels of -400 mg/dl. Blood glucose was followed three times a week, and graft failure was determined by glucose values exceeding 300 mg/dl after at least three days of normoglycemia.
  • Temporary T cell depletion also termed debulking therapy
  • Temporary T cell depletion included a single dose of a mixture of depleting polyclonal anti-CD4 (GK1.5) and anti-CD8 (53.6.72) antibodies (BioXCell), each at 300 ⁇ at the concentration of 1 mg/ml, 3 days prior to transplantation.
  • Subtherapeutic co- stimulation blockade included an equal mixture of anti-LFA-1 and anti-CD 154 monoclonal antibodies (MR-1 and FD441.8, respectively, BioXCell, West Riverside, NH, USA), each at 25 ⁇ 1/1 ⁇ ] ⁇ 1 ⁇ at the concentration of 1.25 mg/ml, one day before transplantation and every three days thereafter.
  • the maximal treatment duration was 40 days.
  • Insulin immunostaining was performed with guinea-pig- anti- swine-insulin, detected by Cy3- donkey- anti-guinea-pig (both 1:200, DakoCytomation, Glostrup, DK); B cell immunostaining was performed with rat-anti-mouse-B220 (1 : 100, eBioscience, San-Diego, CA, USA), detected by DyLight488-goat-anti-rat (l:200,Jackson IR, PA, USA); T cell immunostaining was performed with Armenian-hamster-anti-CD3 (BioLegend, San- Diego, CA, USA), detected by fluorescence isothiocyanate (FITC) -rat- anti- Armenian- hamster (eBioscience), both at 1:50; Treg immunostaining was performed with mouse- anti-mouse-foxp3 (Biolegend), detected by Cy2-donkey-anti-mouse (Jackson IR), both at 1 : 100.
  • FACS analysis Percent CD3+ cells out of circulating CD45+ leukocytes was determined in fresh heparinized whole blood obtained from mouse-tails. Red blood cells (RBC) were lysed using RBC lysis buffer followed by double-staining with FITC-anti- CD3 (BD Biosciences) and APC-anti-CD45 (eBioscience). Each sample contained at least 1 x 106 cells. Percent B cells in DNL were determined in single-cell suspensions of excised lymph nodes. Triple- staining was preformed using phycoerythrin (PE)-anti-CD40, FITC-anti-CD19 and APC-anti-B220 antibodies (all from eBioscience and diluted according to manufacture's recommendation). FACS analysis was carried out using FACS Calibuer (Becton Dickinson). Data was analyzed using CellQuest software.
  • PE phycoerythrin
  • hAAT monotherapy 60 mg/kg from 1 day prior to transplantation
  • both a higher dose 240 mg/kg
  • an extended 10- day pretreatment protocol was tested.
  • hAAT injections were repeated every 3 days in all experiments.
  • a total of n (number in group) 6 mice were grafted under these conditions, including two recipients per modified protocol.
  • n 6 mice were grafted with no added therapy, as control.
  • neither of the three modified hAAT monotherapy protocols delayed islet xenograft rejection day (CT 10,11,12,13,15, 22 and hAAT 11, 12, 13, 14, 15, 24).
  • the extended hAAT protocol is thereby used throughout the following studies.
  • FIG. 2B depicts relative changes in specific transcript numbers. While DLN CD40, IL-6 and IL-10 transcript levels did not increase after xenotransplantation at this time point, CD86 displayed a significant increase from non-grafted mice. In the presence of systemic hAAT, CD40 was reduced by 28.3% on average, CD86 by 21.5%, IL-6 by 40.6% and IL-10 by 32.87% (Figure 2B).
  • Islet xenotransplant survival is extended under hAAT and temporary T cell depletion combination Since monotherapy with hAAT appears to have allowed an uninterrupted xeno- response, we sought to examine the combination of hAAT treatment with a technique for modifying xenoimmunity, namely, temporary T cell depletion.
  • mice a larger percentage of mice (from day 15 onwards) exhibited functional islet xenografts when treated with either a combination of AAT/anti-CD4 or AAT/anti-CD8 (compared to a monotherapy with each of the antibodies or AAT).
  • hAAT and temporary T cell depletion combination results in modified graft site immune infiltration and gene expression profiles
  • hAAT monotherapy resulted in a non-invasive population of mononuclear cells that was located in the region between the renal tissue, capsule and graft, containing Tregs.
  • histological images of islet grafts that lack an immune infiltrate was compared with histological samples collected from untreated xenogenic grafts, as well as xenogenic transplants treated by combination of debulking therapy and hAAT that were either accepted or rejected.
  • hAAT and temporary T cell depletion combination decreases T and B lymphocyte content in xenografts and promotes local foxp3+ Tregs
  • Explanted grafts were analyzed for T and B cell markers, as well as for Tregs immunohistochemistry.
  • representative images from grafts debulking therapy 10 days after rejection, DB/AAT 11 days after rejection and DB/AAT that did not reject.
  • Foxp3-positive Tregs were abundant in the accepted grafts.
  • populations of CD3+ and B220+ cells were reduced in both debulking alone and combined debulking and hAAT, compared to untreated animals (not shown).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Physiology (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Endocrinology (AREA)
  • Nutrition Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Transplantation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/IB2013/051562 2012-02-28 2013-02-27 Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection WO2013128381A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/380,118 US20150010581A1 (en) 2012-02-28 2013-02-27 Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection
EP13755285.7A EP2819698A4 (de) 2012-02-28 2013-02-27 Kombinationstherapie aus alpha-1-antitrypsin und einer temporalen t-zellen-abreicherung zur hemmung einer transplantatabstossung
IL233647A IL233647A0 (en) 2012-02-28 2014-07-14 Combined treatment including alpha-1-antitrypsin and temporary depletion of t-cells to prevent graft rejection
US15/477,379 US10086070B2 (en) 2012-02-28 2017-04-03 Combined therapy of alpha-1-antitrypsin and temporal T-cell depletion for preventing graft rejection

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261603970P 2012-02-28 2012-02-28
US61/603,970 2012-02-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/380,118 A-371-Of-International US20150010581A1 (en) 2012-02-28 2013-02-27 Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection
US15/477,379 Continuation US10086070B2 (en) 2012-02-28 2017-04-03 Combined therapy of alpha-1-antitrypsin and temporal T-cell depletion for preventing graft rejection

Publications (1)

Publication Number Publication Date
WO2013128381A1 true WO2013128381A1 (en) 2013-09-06

Family

ID=49081716

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2013/051562 WO2013128381A1 (en) 2012-02-28 2013-02-27 Combined therapy of alpha-1-antitrypsin and temporal t-cell depletion for preventing graft rejection

Country Status (4)

Country Link
US (2) US20150010581A1 (de)
EP (1) EP2819698A4 (de)
IL (1) IL233647A0 (de)
WO (1) WO2013128381A1 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10980866B2 (en) 2014-09-22 2021-04-20 Hadasit Medical Research Services And Development Ltd. Alpha-1 anti-trypsin for treating liver diseases

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3362088B1 (de) * 2015-10-12 2020-11-25 Institut National de la Sante et de la Recherche Medicale (INSERM) Zur verringerung von cd8-t-zellen fähiger wirkstoff zur behandlung von myokardinfarkt oder akutem myokardinfarkt
US11819215B2 (en) 2018-04-04 2023-11-21 Incumedx Inc. Embolic device with improved neck coverage

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138017A2 (en) * 2007-05-08 2008-11-13 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for modifying t cell immune responses and inflammation
WO2009046015A2 (en) * 2007-09-30 2009-04-09 University Of Florida Research Foundation, Inc. Combination therapies for treating type 1 diabetes

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0631783A1 (de) * 1993-06-03 1995-01-04 Mitsubishi Chemical Corporation Antivirale Kombinationen von 2',3-Di-deoxyribonukleosiden mit 6-benzyl-1-ethoxy-methyl-5-substituierten Uracil-Derivaten
US5968753A (en) * 1994-06-14 1999-10-19 Nexell Therapeutics, Inc. Positive and positive/negative cell selection mediated by peptide release
EP1909810B2 (de) * 2005-06-07 2017-08-23 The Regents of the University of Colorado Hemmer der serinprotease-aktivität und ihre verwendung in verfahren und zusammensetzungen zur behandlung von transplantat-abstossung und förderung des transplantat-überlebens
US9457070B2 (en) * 2005-06-07 2016-10-04 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses of alpha 1-antitrypsin for early intervention in bone marrow transplantation and treatment of graft versus host disease
EP2375892A1 (de) * 2008-12-26 2011-10-19 Baylor Research Institute Ger?t und verfahren f?r die konservierung von pankreasgewebe und inselzellen f?r die transplantation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138017A2 (en) * 2007-05-08 2008-11-13 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for modifying t cell immune responses and inflammation
WO2009046015A2 (en) * 2007-09-30 2009-04-09 University Of Florida Research Foundation, Inc. Combination therapies for treating type 1 diabetes

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BUMGARDNER G.L. ET AL.: "Patterns of Immune Responses Evoked by Allogeneic Hepatocytes: Evidence for independent co-dominant roles for CD4+ and CD8+ T-cell responses in acute rejection", TRANSPLANTATION, vol. 68, no. 4, 27 August 1999 (1999-08-27), pages 555 - 562, XP055162921, Retrieved from the Internet <URL:http://www.centerspan.org/pubs/transplantation/1999/0827/tr169900555p.pdf> [retrieved on 20130619] *
KOULMANDA, M. ET AL.: "Curative and beta cell regenerative effects of alphal-antitrypsin treatment in autoimmune diabetic NOD mice", PROC NATL ACAD SCI USA, vol. 105, no. 42, 21 October 2008 (2008-10-21), pages 16242 - 16247, XP001539157, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2570971/> [retrieved on 20130619] *
See also references of EP2819698A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10980866B2 (en) 2014-09-22 2021-04-20 Hadasit Medical Research Services And Development Ltd. Alpha-1 anti-trypsin for treating liver diseases

Also Published As

Publication number Publication date
EP2819698A1 (de) 2015-01-07
EP2819698A4 (de) 2015-11-11
US10086070B2 (en) 2018-10-02
IL233647A0 (en) 2014-08-31
US20170202961A1 (en) 2017-07-20
US20150010581A1 (en) 2015-01-08

Similar Documents

Publication Publication Date Title
EP2566566B1 (de) Adulte stammzellen/vorläuferzellen sowie stammzellenproteine zur behandlung von augenverletzungen und -erkrankungen
EP2195009B1 (de) Zyklische undekapeptide und derivate als therapien für multiple sklerose
Andres et al. Histology of human tubulo-interstitial nephritis associated with antibodies to renal basement membranes
JP2018512387A (ja) 聴力損傷予防用ペプチド及びそれを含む組成物
EP2978442B1 (de) Alpha 1 antitrypsin zur vorbereitung eines patienten auf ein transplantat
US10086070B2 (en) Combined therapy of alpha-1-antitrypsin and temporal T-cell depletion for preventing graft rejection
US20120309683A1 (en) USE OF PITUITARY ADENYLATE CYCLASE-ACTIVATING POLYPEPTIDE (PACAP) AND PACAP ANALOGS AS ADJUNCTIVE TREATMENTS WITH INHIBITORS OF CALCINEURIN OR INHIBITORS OF THE MAMMALIAN TARGET OF RAPAMYCIN (mTOR) COMPLEXES
AU2018247928B2 (en) C4BP-based compounds for treating immunological diseases
KR20090060290A (ko) 데스아실 그렐린 및 그 유도체를 유효성분으로 포함하는 척수 신경 수복 촉진 치료제
WO2022161484A1 (en) Methods of treating chronic active antibody-mediated rejection using btk inhibitors
WO2014060392A1 (en) Caspase-6 inhibitors for treating t cell activation and/or proliferation disorders
WO2016176493A1 (en) Treatment of medical conditions
EP2773747B1 (de) Tsg-6 protein zur verwendung bei der vorbeugung und behandlung der abstossung einer hornhauttransplantation
US9649356B2 (en) Method of improving transplant function using soluble complement receptor type I (sCR1)
US20050025769A1 (en) Method of suppressing ongoing acute allograft rejection
CA2660260A1 (en) Use of modified interleukin-8 proteins
JP6373963B2 (ja) 移植のためのh因子
KR20190096379A (ko) Ilc2 세포와 연관된 질환을 치료하는 방법
JP6484215B2 (ja) 関節リウマチの治療のためのh因子
CN114887036A (zh) 抗菌肽在制备对乙酰氨基酚诱导的急性肝损伤和/或肝衰竭治疗制剂中的应用
NZ786536A (en) Methods of treating diseases associated with ILC2 cells
Poster Renal Transplantation, Renal Vascular Surgery (II)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13755285

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 233647

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 14380118

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013755285

Country of ref document: EP